Mice
Animals were raised on the Model Animal Platform at ShanghaiTech University. All animal protocols were approved by the Institutional Animal Care and Use Committee of the ShanghaiTech University. The mice were bred from C57BL/6J background and group-housed (3-5 mice per cage) on a 12h/12h light/dark cycle with diet and water freely available. Tg(Egr1-EGFP)GO90Gsat (GENSAT: 4847022) knockin mice, both male and female, aged 8-12 weeks, were bilaterally injected with LV-CRISPR library into the dorsal dentate gyrus (±1.5, -2.0, -2.0). Mice were allowed to recover from surgery for 2-3 weeks before all behavioral tasks. B6.Cg-Tg(Nes-cre)1Kln/J (003771) mice were crossed to Kdm4aflox/flox mice to generate neuron-specific Kdm4a knockout mice (Nes-Cre+/Tg;Kdm4af/f). Nes-Cre mice were kindly gifted by Y. Cang laboratory at ShanghaiTech University.
DNA constructions
Candidate sgRNAs and shRNAs sequences targeted on the genes of interest were designed using E-CRISP51 and CRISPick (https://portals.broadinstitute.org/gpp/public/) online tools. To construct the lentiviral CRISPR plasmids, the stuffer on lenti-CRISPRko or lenti-CRISPRa backbone was replaced with annealed sgRNA oligos using restriction enzyme-based cloning. To generate lentiviral RNAi vectors, the hPGK and PuroR fragments on the pLKO.1 backbone (Addgene, 10878) were replaced with CMV promoter and full-length EYFP respectively using Gibson Assembly, and then the annealed shRNA oligos were inserted to the plasmid using restriction enzyme-based cloning.
To produce the proximity labeling plasmids, KDM4A fragments were PCR amplified from the cDNA library of human 293T cell line and fused to the N-terminal of PafA with a 3xFlag-tag. KDM4A-PafA fragments were subcloned into the lentiviral plasmids using Gibson Assembly. A nuclear localization signal (NLS) was fused to the N-terminal of PafA, as a control bait. The N-terminal of Bio-PupE, the substrates of PafA, was fused to a 3x NLS tag for the localization in the nuclei. IRES fragments and EGFP or mCherry fragments were inserted into the C-terminal of KDM4A-PafA or 3x NLS-PafA using Gibson Assembly. All plasmids were verified by Sanger sequencing.
Generation of the sgRNA library for the screen
The mouse histone methylation CRISPR sgRNA library was designed by selecting ten sgRNAs targeting each gene of interest (117 genes encoding histone methylation modifiers were obtained from the EpiFactor Database52,53), along with an additional 15 non-targeting negative control sgRNAs. The oligonucleotide pool was synthesized by CustomArray, Inc.
The pooled library sgRNA fragments, which contained homology arms, were inserted into the BsmB1-digested lentiCRISPRv2-mCherry plasmids using Gibson Assembly Kit (NEB). The ligation products were desalted and transformed into 20 μl of DH5α competent cells (Weidi, DE1001) using an electroporation apparatus (Bio-Rad MicroPulser). After electroporation, the cells were recovered by adding 980 μl of pre-warmed 37℃ SOC medium (Weidi, CM1014L) and incubating for 1 h at 37℃ with shaking at 250 rpm. Transformants were plated onto five 24.5 x 24.5 cm LB agar plates and incubated for 14-15 h at 30℃. The colonies were collected and subjected to plasmid DNA extraction using an endotoxin-free plasmid DNA isolation kit.
To examine the integrity and proper representation of the sgRNAs in the library plasmids, the plasmid DNAs were used for PCR amplification of regions containing the sgRNA fragment. The PCR products were purified and determined by Next Generation Sequencing (NGS).
Cell culture and transfection
The HEK293T, HEK293FT, U2-OS and Neuro-2a cell lines were maintained in high-glucose DMEM medium supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin at 37℃ with 5% CO2. All cells were subcultured every 3-4 days when they reached 80% confluency. The transfection of HEK293T, HEK293FT cells was performed using polyethylenimine (PEI). The transfection of U2-OS and Neuro-2a cells was performed using EZ Trans Reagents (Life-iLab).
Lentivirus production
Lentivirus was generated by transfected HEK293-FT cells in three 10-cm cell culture dishes with lentiviral transfer vectors, packaging vector psPAX2 and envelop vector pVSV-G using polyethylenimine (PEI). Media containing viral particles were harvested from cell culture at 48 and 72 h after transfection. The collected media were passed through a 0.45 μm filter to remove cell debris, and then the filtered media were centrifuged in a Beckman SW-28 rotor for 2 h at 25,000 rpm at 4℃, and 5 ml of 20% sucrose solution was added to the bottom of the centrifuge tube before centrifugation. The concentrated lentivirus was resuspended in 20 μl ice-cold D-PBS and stored at -80℃.
Stereotaxic surgery and viral injection
For delivery of virus into the DG, mice were anesthetized using isoflurane (4% for induction and 1.5% for maintenance) and mounted in a stereotaxic apparatus. The fur on mouse head was trimmed to expose the skin, which was then sterilized using 70% ethanol followed by 10% iodophor solution. A 1-cm skin incision was performed to find the Bregma point on the skull, and the placement of injection points was determined in relation to Bregma. Burr holes were drilled using a sterile hand drill to expose the brain. After craniotomy, 1 μl of lentivirus or 350 nl of AAV virus was slowly injected (50 nl/min) into the bilateral DG (coordinates from Bregma: -2 mm AP, ±1.5 mm ML, -2 mm DV) using a microsyringe pump. The syringe was left in tissue for 10 minutes after each injection to allow the viral particles to diffuse. After viral injection, the surgical incision was closed with sutures and the mice were then placed on a heating pad for recovery. Behavioral tasks were conducted at 2 weeks after the surgeries.
In this paper, LV-U6-shCtrl-CMV-EYFP (titer, 2.42 x 109 TU/ml), LV-U6-shKdm4a-CMV-EYFP (titer, 1.42 x 109 TU/ml), LV-U6-sgCtrl-EF1α-nls-Cas9-P2A-EGFP (titer, 2.64 x 108 TU/ml), LV-U6-sgKdm4a-EF1α-nls-Cas9-P2A-EGFP (titer, 2.87 x 108 TU/ml) were packaged by OBiO Technology (Shanghai) Co., Ltd. AAV2/9-CaMKIIα-hM3Dq-mCherry (titer, 2.67 x 1012 genomic copies per ml), AAV2/9-CaMKIIα-mCherry (titer, 2.59 x 1012 genomic copies per ml) were purchased from OBiO Technology (Shanghai) Co., Ltd.
Fluorescence activated cell sorting
Fluorescence activated cell sorting was performed as previously described15,27. Mice were anesthetized using overdosed isoflurane. Mouse brain was perfused with 20 ml ice-cold perfusion buffer (115 mM Choline chloride, 2.5 mM KCl, 1.25 mM NaH2PO4, 26 mM NaHCO3, 10 mM glucose, 8 mM MgSO4, 1 mM Sodium L-ascorbate and 3 mM Sodium pyruvate, pH 7.4) that pre-bubbled with 5% CO2/95% O2 on ice for 30 min. After perfusion, mouse brain was dissected and sectioned using pre-chilled blades and coronal brain matrices for rodents (1 mm thick). Brain sections were immersed in pre-chilled modified EBSS buffer (116 mM NaCl, 5.4 mM KCl, 1 mM NaH2PO4, 26 mM NaHCO3, 1.5 mM CaCl2, 1 mM MgSO4, 0.5 mM EDTA, 25 mM glucose and 1 mM L-Cysteine, pH 7.4) in a 6-cm cell culture dish. The dentate gyrus was microdissected and rinsed with fresh modified EBSS buffer. Tissue pieces were transferred to a new 15-ml Falcon tube containing 2 ml of modified EBSS buffer and centrifuged at 300 xg for 5 min at 4℃. After centrifugation, the tissue pieces were gently resuspended with 3 ml of digestion buffer (modified EBSS buffer supplemented with 20 units/ml papain and 0.005% DNase I) and then incubated in a water bath at 37℃ for 45 min with 5% CO2/95% O2. After papain-based enzymic dissociation, tissues were centrifuged at 300 g for 5 min at 4℃ and the pellet was resuspended with 1 ml of stop solution (modified EBSS buffer supplemented with 1 mg/ml Trypsin inhibitor, 1 mg/ml BSA and 0.005% DNase I) to quench papain. The tissues were gently triturated through fire-polished P1000 pipette tips, then fire-polished P200 pipette tips. To remove myelin, cell suspensions were centrifuged at 300 xg for 5 min at 4℃, resuspended in 1.5 ml of stop solution supplemented with 0.9 M sucrose, and centrifuged at 600 xg for 10 min at 4℃. The cell pellet was then resuspended with 0.5 ml stop solution and passed through a 100-μm cell strainer to remove debris. 10 μg/ml DAPI was added and incubated on ice in the dark for 5 min before cell sorting. Live cells were sorted using the BD FACS Aria III instrument into 1.5-ml EP tubes containing stop solution. Before sorting, unstained controls and single-color stained controls were used to validate the gating strategies. Dead cells were excluded by staining with DAPI.
For genomic DNA isolation, cells were collected by spinning at 2,000 xg for 10 min at 4℃. The pellets were then snap frozen and stored at -80℃ until downstream analysis. For RNA extraction, cells were sorted directly into lysis buffer and immediately snap frozen until reverse transcription.
Preparation of samples for NGS
For the preparation of sequencing libraries, genomic DNA was extracted using the QIAamp DNA Mini Kit (QIGEN, 51304) with carrier RNA (Poly rA) (QIAGEN, 1068337). The integrated sgRNA sequences were PCR amplified and barcoded using the Q5 Hot Start HiFi DNA Polymerase (NEB, M0515). The libraries were then purified and sequenced using the Illumina HiSeq X system.
CRISPR screen analysis
For the MAGeCK-Flute54,55 analysis of the CRISPR screen, two fastq files, named GFP-mCherry+ and GFP+mCherry+, were used as input to a Python-based MAGeCK-Flute platform to calculate the read counts and fold changes for GFP+mCherry+ versus GFP-mCherry+ to score and rank. The read counts for each sgRNA were normalized as follow:
Plasmid fastq file were used as a control for MAGeCK-Flute analysis. P values and gene rankings generated from MAGeCK-Flute analysis were used for identification of candidate genes.
T7E1 mismatch detection assay
The sgRNAs targeting the mouse Kdm4a were designed using the E-CRISP51 sgRNA designing tool. Kdm4a-targeting sgRNAs were cloned into lentiCRISPRv2-EGFP vectors. Neuro-2a cells were transiently transfected with the plasmids using EZ Trans reagent. EGFP+ cells were sorted through flow cytometry. Then, genomic DNAs were extracted and used for PCR amplification of the regions containing sgRNA targeting site. The PCR products were purified and subjected to denaturing and reannealing. The hybridization reaction generates mismatches that are cleaved by the T7 endonuclease 1 (T7E1). Half of the sample was kept as an undigested control. 1 μl of T7 endonuclease 1 was added to the remaining sample and incubated for 15 min at 37℃. The digested and undigested hybridized DNA samples were resolved by 2% agarose gel electrophoresis.
TIDE analysis
The genomic DNA was extracted from CRISPR-editing cells, as well as an unedited control. The genomic regions containing sgRNA-targeting site were PCR amplified. The PCR products were purified and used for Sanger sequencing. The sequencing files for the edited and control samples, and 20-nt guide sequence upstream of PAM, were submitted to the TIDE56 (Tracking of Indels by DEcomposition) or ICE57 (Inference of CRISPR Edits) web tools for identification of the predominant types of indels and quantification of the editing efficacy.
Mouse neuron culture
Embryonic cortices from C57BL/6J mice were dissected at E16.5 and dissociated with 0.25% Trypsin and 0.04% DNase I in the dissection medium (1x HBSS, 10 mM HEPES, 11 mg/ml sodium pyruvate, 0.1% glucose) at 37℃ for 15 min. Digested cortices were centrifuged and gently triturated through fire-polished P1000 pipette with plating medium (Neurobasal medium, 10% FBS, 1% GlutaMAX Supplement, 1% Pen/Srep). Neurons were plated on cell culture dishes pre-coated with coating solution (20 μg/mL poly-D-lysine, 4 μg/mL laminin) overnight. Neurons were diluted and plated at a density of 106 cells/dish on 3.5-cm dishes. Neurons were grown in maintenance medium (Neurobasal medium, 2% B-27 Supplement, 1% GlutaMAX Supplement, 1%Pen/Srep) at 37℃ with 5% CO2 for 12-14 days. Half of the maintenance medium was changed every 3-4 days.
For virus infection experiments, the lentivirus was added at DIV3 and the infected neurons were collected at DIV10 for RNA isolation and RT-qPCR assay. For high-potassium chloride (KCl) depolarization experiments, DIV14 neurons were incubated with 1/3 volume of depolarization solution (170 mM KCl, 10 mM HEPES, pH7.4, 1 mM MgCl2, 2 mM CaCl2) into culture medium (final 55 mM KCl) for 1 h to achieve neuronal activation. Neurons were collected after 0 h or 1 h KCl-induced depolarization.
RNA-seq analysis
Fluorescent immunostaining
Cultured cells were fixed using 4% paraformaldehyde (PFA) in PBS at 4℃ for 10 min. Mice were anesthetized using an overdose of isoflurane. The mouse brain was perfused and fixed with 4% PFA in PBS at 4℃ overnight. Samples were sectioned coronally (40 μm-thickness) on a Leica Vibrating blade microtome. Fixed cells and brain slices were blocked and permeabilized with blocking buffer (1% bovine serum albumin, 0.3% Triton X-100 in PBS, pH 7.4) at 4℃ for 1 h, followed by incubation with primary antibodies diluted in the blocking buffer at 4℃ overnight with gentle shaking. After washing, secondary antibodies diluted in the blocking buffer were applied to the samples at room temperature for 2 h. Nuclei were stained by incubation with 0.5 μg/mL 4,6-diamidine-2-phenylindole (DAPI) in PBS at room temperature for 10 min. The samples were washed thrice with PBS for 15 min each time. The stained samples were mounted with anti-fade mounting medium and used for confocal microscopy.
RNA extraction and RT-qPCR
Cultured cells or mouse brain tissues that were dissected were homogenized using 0.2 ml of TRIzol reagent (Invitrogen) and incubated at room temperature for 5 min. Add 40 μl chloroform to the cell lysate and vortex the samples vigorously. RNA remained in the aqueous phase after centrifugation at 12,000 xg for 15 min at 4℃. The upper aqueous phase that contained RNA was collected for precipitation by adding 0.1 ml of isopropyl alcohol. Samples were mixed and incubated at room temperature for 10 min and centrifuged at 12,000 xg for 10 min at 4℃. The RNA pellet was washed twice with 0.1 ml of 75% ethanol and centrifuged at 12,000 xg for 5 min at 4℃. The RNA pellet was air-dried for 5 min and dissolved in 20 μl of DEPC-treated H2O. The concentration of RNA was determined by a spectrophotometer at 260 and 280 nm.
Reverse transcription was performed using the HiScript III 1st Strand cDNA Synthesis Kit (Vazyme, R312-01). The cDNAs were used for SYBR Geen (Bimake, B21202)-based quantitative real-time PCR to determine the relative expression levels of genes of interest using the ΔΔCt quantification method.
RNA-seq analysis
For the preparation of sequencing samples, lentivirus LV-U6-shKdm4a-CMV-EYFP or LV-U6-shCtrl-CMV-EYFP were delivered into the adult male C57BL/6J mouse DG through stereotaxic injection. After 14 days of recovery, mice were anesthetized and perfused with ice-cold PBS. The DG was microdissected and dissociated with papain-based enzymatic digestion. 2,000 EYFP+ cells were sorted using the BD FACS Aria III instrument into a 200-μl PCR tube containing lysis buffer and RNase inhibitor. After sorting, the tubes were immediately snap-frozen. Total RNA was reverse transcribed using the SMART-Seq HT Kit (Clontech, 634437), and cDNAs were used to produce the sequencing library. The libraries were purified and sequenced using the Illumina NovaSeq 6000 system.
For RNA-seq data analysis, raw reads were filtered using Seqtk (v.1.4) to remove adapters and low-quality regions, and mapped to the GRCm38 genome using Hista258 (v.2.0.4). Reads on each gene were counted by Stringtie59 (v.1.3.0). Differential expressed genes (DEGs) were identified using edgeR. Gene ontology analysis was performed by online GESA60 tools using all significant DEGs (P <0.05).
Western blot
The proteins in cell lysates were resolved by 6%, 10% or 12% SDS-polyacrylamide gel electrophoresis, depending on the molecular weight of the proteins of interest, and transferred to a polyvinylidene difluoride membrane. The membrane was blocked with 5% skimmed milk in TBST buffer (20 mM Tris, 150 mM NaCl, 0.1% Tween-20, pH 7.6) for 1 h. The membrane was washed three times with TBST buffer and incubated with primary antibodies diluted in the TBST buffer with 5% bovine serum albumin (BSA) overnight with gentle shaking at 4℃. After washing, HRP-conjugated secondary antibodies diluted in TBST buffer with 5% BSA were applied to the membrane at room temperature for 1 h. The membrane was then washed three times with TBST buffer for 15 min. The signals on the membrane were detected in the ChemiDoc imaging system (Bio-Rad).
Chromatin Immunoprecipitation
For each sample, 20 μl of Protein-G magnetic beads were washed twice with beads binding buffer (1x PBS, pH 7.4, 0.2% Tween-20). The washed beads were incubated with 2 μg of antibodies in 200 μl of beads binding buffer overnight at 4℃ on a rotator. Bead-antibody complexes were washed thrice with beads binding buffer and then washed twice with dilution buffer (0.01% SDS, 1.1% Triton X-100, 1.2 mM EDTA, pH 8.0, 16.7 mM Tris, pH 8.1, 167 mM NaCl). Washed bead-antibody complexes were added with 50 μl dilution buffer and stored at 4℃ until cells were ready.
Dissected mouse brain tissues were dissociated using a homogenizer with 1 ml of ice-cold PBS supplemented with 1x Protease inhibitors cocktail. The lysates were crosslinked with 27 μl of 37% paraformaldehyde (final 1% PFA) for 15 min at room temperature on a rotator. 83.3 μl of 2 M glycine were added (final 150 mM glycine) and incubated on a rotator for 10 min to quench the crosslink reaction. Cells were collected by centrifugation at 2,000 xg for 10 min at 4℃. The cell pellets were washed and resuspended with 300 μl cell lysis buffer (50 mM Tris, pH 8.0, 140 mM NaCl, 1 mM EDTA, pH 8.0, 10% glycerol, 0.5% NP-40, 0.25% Triton X-100 supplemented with 1x Protease inhibitors cocktail) for a 20 min of incubation on ice. Nuclei were collected by centrifugation at 2,000 xg for 5 min at 4℃. The nuclear pellets were resuspended with 300 μl nuclear lysis buffer (10 mM Tris, pH 8.0, 1 mM EDTA, pH 8.0, 0.5 mM EGTA, pH 8.0, 0.5% SDS supplemented with 1x Protease inhibitors cocktail). After incubation for 20 min on ice, lysates were sonicated at 9 cycles of 50% power 30 s on/off treatment at 4℃ to fragment the chromatin. The samples were centrifuged at 1,6000 xg for 10 min at 4℃ to remove insoluble debris. The supernatants were collected and stored on ice. Part of the supernatants were kept as input and stored at -80℃. Next, the remaining supernatants were added and incubated with 50 μl bead-antibody complexes overnight on a rotator at 4℃. Then, the beads were washed five times with wash buffer (100 mM Tris, pH 8.0, 500 mM LiCl, 1% NP-40, 1% Sodium deoxycholate) and washed once with TE buffer (50 mM Tris, pH 8.0, 10 mM EDTA). The washed beads were then resuspended in 170 μl elution buffer (50 mM Tris, pH 8.0, 10 mM EDTA, 1% SDS) and incubated for 20 min at 65℃. The eluted samples and input samples were both incubated in the elution buffer overnight at 65℃ for decrosslinking. Then, 1 μl 10 mg/ml RNase A was added in each sample and incubated at 37 ℃ for 1 h to remove RNA. 1 μl 20 mg/ml Proteinase K was added and incubated at 55℃ for 2 h to digest protein. Next, the input and ChIP DNAs were extracted using phenol/chloroform/isoamyl alcohol (25:24:1). The purified DNAs were measured on a Nanodrop and then used for real-time qPCR analysis.
Proximity labeling assay
In proximity labeling assay, the HEK293T cells transfected with KDM4A-PafA and 3x NLS-Bio-PupE (KDM4A-PUPIT) plasmids were treated with 4 μM biotin for a 24 h of incubation. Control cells were transfected with 3xNLS-PafA and 3x NLS-Bio-PupE (CTRL-PUPIT) plasmids. The nuclear pupylated proteins were extracted with RIPA lysis buffer (50 mM Tris, pH 7.4, 150 mM NaCl, 1% Triton X-100, 1% Sodium deoxycholate, 0.1% SDS, 5 mM EDTA supplemented with 1x Protease inhibitors cocktail and 1x Phosphatase inhibitors cocktail) and then used for streptavidin immunoprecipitation. The enriched proteins were incubated at 95℃ in 2x Laemmli loading buffer for 10 min and then subjected to western blot or mass spectrometry analysis.
Mass spectrometry analysis
For the preparation of MS samples, the enriched proteins were first resolved by 10% SDS-PAGE, and electrophoresing for 2 cm in the gel. After electrophoresis, the gel was washed thrice with ddH2O for 15 min. For each sample, the entire region containing proteins were excised and cut into 1 mm slices, and then collected in a 1.5-ml tube. After washing with ddH2O, the gel slices were incubated with 0.5 ml acetonitrile (ACN) to dry up the gel pieces at 37℃ for 10 min at 500 rpm, and repeat this step once. Completely dehydrate the gel slices using a vacuum centrifuge for 10 min. 0.5 ml of 10 mM DTT in 25 mM NH4HCO3 solution were added in the gel slices and incubated at 65℃ for 1 h at 500 rpm. The supernatants were removed, and 0.4 ml of 50 mM iodoacetamide (IAM) in 25 mM NH4HCO3 solution was then added for alkylation, and incubated in the dark at room temperature for 45 min. The supernatants were replaced by 25 mM NH4HCO3 solution, and incubated at room temperature for 10 min on a rotator. After removal of supernatants, same volume of 25 mM NH4HCO3 solution and ACN were added in the gel slices and incubated at 37℃ for 5 min at 500 rpm. The supernatants were replaced by 0.5 ml of ACN, and incubated at 37℃ for 10 min at 500 rpm. Completely dehydrate the gel slices using a vacuum centrifuge for 10 min. For each sample, 3 μl of Trypsin (Promega, v5113) and 0.5 ml 25 mM NH4HCO3 solution were added in gel slices and then digested at 37℃ overnight at 500 rpm. On the following day, the digested proteins were centrifuged at 3,000 xg at room temperature for 3 min. For each sample, 400 μl of 50% ACN, 0.5% formic acid (FA) solution was added and incubated at 37℃ for 10 min at 500 rpm. The supernatants were transferred into a new 1.5-ml tube, and repeat this step once with 200 μl of 50% ACN, 0.5% FA solution. Completely dehydrate the combined supernatants using a vacuum centrifuge for 3-4 h. After dehydration, 50 μl of 0.1% FA solution was added to dissolve the peptides. To remove the salt from sample, Pierce C18 tips (Thermo, SP301) were washed twice with 200 μl buffer-B (50% ACN, 0.1% FA), and centrifuged at 500 xg for 3 min. The tips were then washed twice with 100 μl buffer-A (0.1% FA), and centrifuged at 500 xg for 3 min. The washed tip was transferred to a new 1.5-ml tube. Then, the solution containing peptides was injected into the Pierce C18 tips and centrifuged at 500 xg for 3 min, and then collected the follow-through. The follow-through was re-injected into the tips and centrifuged again. The tips containing peptides were washed with 100 μl buffer-A and then transferred to a new 1.5-ml tube. for elution of peptides, 50 μl of buffer-B was added into the tips to elute the peptides that bind to the membrane in the tip, and centrifuged at 500 xg for 3 min. This step was repeated once. The eluted peptides were completely dehydrated using a vacuum centrifuge for 1 h. The sample was then dissolved in 10 μl of ACN solution, and 3 μl of the solution was injected into the LC-MS/MS instruments. The peptides were identified and quantified using MaxQuant61 and Perseus62 platforms.
Co-Immunoprecipitation
About 107 HEK293T cells that stably co-transfected with Flag-KDM4A and HA-YTHDC2 plasmids were used to detect the interaction between KDM4A and YTHDC2. Cells were incubated with 4 ml crosslink solution (1 mM DSP in PBS, pH 7.2) in 10-cm cell cultured dishes at room temperature for 30 min. Same volume of stop solution (40 mM Tris, pH 7.5 in PBS) were added (final 20 mM Tris) and incubated at room temperature for 15 min to quench the crosslink reaction. Cells were collected and lysed with Co-IP lysis buffer (50 mM Tris, pH 7.4, 150 mM NaCl, 1% Triton X-100, 1% Sodium deoxycholate, 0.1% SDS, 5 mM EDTA and 1x Protease inhibitor cocktail) for 20 min on ice. The lysate was centrifuged to remove cell debris at 15,000 xg for 10 min at 4℃. The supernatants were then collected and kept on ice. Protein concentration was measured using BCA Kit (Beyotime, P0010S). Part of the protein samples were kept as input and stored at -80℃.
For each protein immunoprecipitation, 30 μl pre-washed protein G magnetic beads incubated with 3 μg of antibodies (anti-Flag, anti-HA and anti-IgG respectively) overnight on a rotator at 4℃. Then, the washed antibody-beads complexes were added to each remaining supernatant, followed by incubation overnight at 4℃ on a rotator. The beads were collected and washed five times with Co-IP lysis buffer. The enriched proteins were eluted with 3x Laemmli loading buffer. The input and IP samples were incubated at 95℃ in Laemmli loading buffer for 10 min and then used for western blot analysis.
Nuclear run-on (NRO) assay
NRO assay was performed as previously described with some modifications63. For each replicate, 5x106 Neuro-2a cells were collected and washed twice with ice-cold PBS. Cells were collected by centrifugation and the pellet was resuspended in 1 ml of cell lysis buffer (10 mM Tris-HCl, pH 7.4, 10 mM NaCl, 3 mM MgCl2, 0.5% NP-40 supplemented with 40 U/ml RNase inhibitor and 1x Protease inhibitors cocktail). After a 5 min incubation on ice, the lysates were centrifuged at 300 xg for 5 min at 4℃ to collect nuclei. The nuclear pellet was then resuspended in 0.5 ml nuclear lysis buffer (10 mM Tris-HCl, pH 7.4, 3 mM CaCl2, 2 mM MgCl2, 0.5% NP-40, 10% glycerol supplemented with 40 U/ml RNase inhibitor and 1x Protease inhibitors cocktail). The nuclear lysates were then centrifuged and the pellet were resuspended in 40 μl nuclear storage buffer (50 mM Tris-HCl, pH 8.3, 0.1 mM EDTA, 5 mM MgCl2 and 40% glycerol) and kept on ice. For each sample, 60 μl of run-on buffer (10 mM Tris-HCl, pH 8.0, 1 mM DTT, 5 mM MgCl2, 0.3 M KCl, 1% Sarkosyl, 100 U/ml RNase inhibitor supplemented with 1 mM ATP/GTP/CTP, 0.5 mM UTP and Br-UTP) was added in the lysates and mixed gently. The reaction mix was incubated in a water-bath at 30℃ for 30 min. After run-on reaction, 12 μl of 2x DNase I was added for another 5 min of incubation at 30℃ to remove DNA. Protein digestion buffer (20 mM Tris-HCl, pH 7.4, 2% SDS, 10 mM EDTA supplemented with 200 μg/ml Proteinase K) of the same volume was added in the run-on sample and incubated at 55℃ for 1 h to digest protein. RNAs were purified from samples using phenol/chloroform extraction and eluted in 100 μl of DEPC-treated H2O. For nascent RNA immunoprecipitation, 30 μl of Protein-G magnetic beads were washed thrice with PBST buffer (0.1% Tween-20 in PBS) and resuspended in 30 μl PBST buffer. Beads were incubated with 2 μg of Mouse anti-BrdU antibody (Sigma, B2531-.2ML) at room temperature for 10 min. Blocking buffer (0.1% PVP, 1 mg/ml BSA in PBST buffer) was added in the bead-antibody complexes and incubated for another 30 min of incubation. Next, bead-antibody complexes were washed twice with PBSTR (PBST buffer supplemented with 40 U/ml RNase inhibitor) and resuspended in 100 μl PBSTR. RNAs extracted from run-on reaction were incubated at 65℃ for 5 min to open their secondary structure, and then added in the bead-antibody complexes. After 30 min of incubation, beads were collected and washed thrice with PBSTR. Then, the enriched nascent RNAs were eluted using 0.5 ml of TRIzol reagent and isolated by phenol/chloroform extraction. 10 μl of the purified nascent RNAs were used for reverse transcription and RT-qPCR analysis.
RNA Immunoprecipitation (RIP)
RIP was performed as previously described64. Dissected mouse brain tissues were dissociated using a homogenizer in 1 ml RIP buffer (50 mM Tris, pH 7.5, 150 mM NaCl, 1 mM EDTA, pH 8.0, 1 mM DTT, 0.5% NP-40 supplemented with 40 U/ml RNase inhibitor and 1x Protease inhibitors cocktail) for 20 min on ice with gentle shanking. The lysates were centrifuged to remove cell debris at 16,000 xg at 4℃ for 10 min. The supernatants were then collected and kept on ice. Part of the supernatants were kept as input and stored at -80℃. The remaining supernatants were incubated with 4 μg of anti-Ythdc2 antibody or control IgG at 4℃ overnight on a rotator. Pre-washed Protein-G magnetic beads were added in each antibody-lysate complex, followed by 3 h of incubation. Then, the beads-antibody-RNA complexes were collected and washed three times with RIP lysis buffer at 4℃. The enriched RIP-RNAs were eluted using 0.5 ml TRIzol reagent for RNA isolation. RIP-RNA and Input RNA samples were both incubated with TRIzol reagent on ice for 5 min. Add 100 μl chloroform in RNA samples and vortex samples vigorously, followed by 5 min of incubation. RNA remained in the aqueous phase after centrifugation at 15,000 xg for 10 min at 4℃. The upper aqueous phase that contained RNA was collected for precipitation by adding 20 μl 3 M Sodium acetate, pH 5.2, 1 μl 20 mg/ml Glycogen and 500 μl 100% ethanol. Samples were mixed and incubated overnight at -80℃. Samples were then centrifuged at 15,000 xg for 25 min at 4℃. The RNA pellet was washed with 0.2 ml 75% ethanol and centrifuged at 15,000 xg for 15 min at 4℃. The RNA pellet was air-dried for 5 min and dissolved in 20 μl DEPC-treated H2O. 10 μl of each RNA sample were used for reverse transcription and RT-qPCR analysis.
m6A Immunoprecipitation
m6A-RIP was performed as previously described with some modifications65. For each sample, 20 μg of total RNA that was harvested from the mouse brain was incubated with RNA fragmentation buffer (10 mM Tris-HCl, pH 7.5, 10 mM ZnCl2 in DEPC-treated H2O) in a 200-μl PCR tube at 94℃ for 4 min to fragment RNAs into about 200-nt-long fragments. After incubation, the samples were transferred on ice immediately. 2 μl of 0.5 M EDTA solution was added to the samples to stop the fragmentation. The purified fragmented RNAs were eluted in 100 μl DEPC-treated H2O. Part of the RNA samples were kept as input and stored at -80℃. The remaining fragmented RNAs were diluted with 1x IP buffer (10 mM Tris-HCl, pH 7.5, 150 mM NaCl, 0.1% NP-40 in DEPC-treated H2O supplemented with 40 U/ml RNase inhibitor and 2 mM RVC). 5 μl of rabbit anti-m6A antibody or control IgG were added to the RNA samples, which were then incubated for 2 h at 4℃ on a rotator. 30 μl of Protein-G magnetic beads were washed twice with 1x IP buffer and then blocked with 0.5 mg/ml BSA in 1x IP buffer for 2 h at 4℃. The beads were then washed twice with 1x IP buffer and added to the antibody-RNA complexes for another 4 h of incubation at 4℃. The beads-antibody-RNA complexes were collected and washed three times with 1x IP buffer at 4℃. The enriched m6A-RIP-RNAs were eluted using 0.5 ml TRIzol reagent for RNA isolation. The purified m6A RNA and input RNA samples were dissolved in 20 μl DEPC-treated H2O. 10 μl of each RNA sample were used for reverse transcription and RT-qPCR analysis.
Dual luciferase reporter assay
Dual luciferase reporter assay was performed as previously described66. To generate Trpm7 WT renilla luciferase reporter vector, the fragment of exon 12 (from mouse Trpm7 CDS) containing the predicted m6A sites was PCR amplified from Nuro-2a cell cDNA library and cloned into the psiCheck2 vector between the restriction enzyme sites Not I and Xho I. The putative two m6A sites on Trpm7 exon 12 were then replaced by Thymine (T) using site-directed overlapping PCR mutagenesis to generate Trpm7 Mut reporter vector. Full-length Ythdc2 CDS fragments were amplified by PCR and cloned into pcDNA3.1 vector using Gibson Assembly.
The Neuro-2a cells were seeded in a 96-well plate and maintained until they reached 80% confluency. 100 ng of luciferase reporter vectors (Trpm7 WT or Mut) and 100 ng of Ythdc2 CDS or mock vectors were co-transfected using EZ Trans Reagent (Life-iLab). After 36 hrs of transfection, the cells were lysed with 100 μl of lysis buffer (Beyotime) for 15 min at room temperature with gentle shanking. The lysates were centrifugated at 12,000 xg for 5 min to remove cell debris. 80 μl of the supernatants were used for measurement of firefly (Fluc) and renilla (Rluc) luciferase activities with Dual Luciferase Reporter Gene Assay Kit II (Beyotime, RG029S) in the SpectraMax i3 microplate reader. The relative luciferase activity was calculated by dividing Fluc by Rluc and normalized to control. For measuring the Rluc and Fluc mRNA levels, the cells were lysed with TRIzol reagent and total RNAs were purified for RT-qPCR analysis. The relative luciferase mRNA changes were calculated as described above.
Generation of Kdm4afl/fl mice
Kdm4afl/fl transgenic mice were generated by using the CRISPR/Cas9 system. Exon 3 of mouse Kdm4a was targeted by inserting a loxP site in intron 2-3 and a loxP site in intron 3-4. A DNA donor containing targeted exon flanked by two loxP sites and the CRISPR/Cas9 systems were microinjected into the C57BL/6J mouse fertilized eggs. Edited eggs were transplanted to obtain positive offspring, which were determined using PCR amplification and Sanger sequencing. Chimeric mice were then bred to wild-type C57BL/6J mouse to establish a stable transgenic mouse line carrying the loxP sites.
Behavioral tests
Open field
Mice were placed in the center of a 46 x 46 cm box and allowed to freely explore for 10 min after 3 min of habituation. Videos were recorded and analyzed by TSE apparatus and software. The 20 x 20 cm region of the box was defined as central zone. The time in center and the total distance traveled were analyzed for evaluating the mouse locomotor activity.
Contextual fear conditioning
Mice were placed in the footshock chamber (20 x 20 cm) with stainless steel grid floor, and habituated for 3 min. After habituation, mice received a 1 s foot shock of 0.6 mA, followed by 30 s of delay. The next day (24 h later), mice were returned to the footshock chamber without shocks for 3 min of exploration (recall). Mouse activities were analyzed by TSE apparatus and software. The percentage of freezing during recall phase was analyzed for determining the ability of fear memory formation in mouse.
Fear memory extinction
Mice were subjected to contextual fear conditioning and then returned to the home cage after recall session. Two weeks later, mice were re-expose to the same footshock chamber without receiving foot shock for 3 min. After exploration, mice were returned to their home cage. This procedure was repeated for 6 days. The memory extinction curve was analyzed for access the ability of active forgetting in mouse.
Contextual fear discrimination
To test the ability of pattern separation, mice were subjected to a contextual fear discrimination task. Briefly, mice were placed daily in two similar contexts: context A and context B. The footshock chamber (context A) and no-shock chamber (context B) shared the same stainless steel grid floor. Context A was a square box and illuminated with white light. Context B was a circular box and illuminated with yellow light, and 0.5% acetic acid odor were delivered below the floor. 75% ethanol were used to clean grid floor before the mouse was placed in the context. The mice were first placed in context A and received a 1s foot shock of 0.4 mA after 3 min of exploration on day 0. From day 1 to day 4, mice were randomly placed in either context A or context B for 3 min, and then returned to their home cage. 2 h after the first exploration, mice were placed in the other context. The freezing levels in context A and context B each day were calculated and used to determine the discrimination index: (FreezingA - FreezingB) / (FreezingA + FreezingB).
Elevated plus maze
The elevated plus maze consists of two open arms and two closed arms (30 cm long and 6.5 cm wide), both of which are 55 cm above the floor. Mice were placed in the central area and allowed to freely explore the maze for 5 min. Mouse activities were recorded by a camera above the maze and analyzed using a mouse tracking system (Noldus). The time that mice spent in the open arms and closed arms, and a percentage of being in open arms, were calculated to access anxiety-like behavior.
Statistics
Statistic analysis was performed using GraphPad Prism (v.9.0.0) software. Data were represented as mean ± s.e.m. Box or violin plots were represented as the mean, interquartile range and the minimum and maximum. Statistical significance was determined by unpaired or paired two-tailed Student’s t-test, one or two-way ANOVA. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
References
51. Heigwer, F., Kerr, G. & Boutros, M. E-CRISP: fast CRISPR target site identification. Nat. Methods 11, 122–123 (2014).
52. Medvedeva, Y. A. et al. EpiFactors: a comprehensive database of human epigenetic factors and complexes. Database 2015, bav067 (2015).
53. Marakulina, D. et al. EpiFactors 2022: expansion and enhancement of a curated database of human epigenetic factors and complexes. Nucleic Acids Res. 51, D564–D570 (2023).
54. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).
55. Wang, B. et al. Integrative analysis of pooled CRISPR genetic screens using MAGeCKFlute. Nat. Protoc. 14, 756–780 (2019).
56. Brinkman, E. K., Chen, T., Amendola, M. & van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168–e168 (2014).
57. Conant, D. et al. Inference of CRISPR Edits from Sanger Trace Data. Cris. J. 5, 123–130 (2022).
58. Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019).
59. Pertea, M. et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015).
60. Subramanian, A. et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. 102, 15545–15550 (2005).
61. Tyanova, S., Temu, T. & Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 11, 2301–2319 (2016).
62. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).
63. Roberts, T. C. et al. Quantification of nascent transcription by bromouridine immunocapture nuclear run-on RT-qPCR. Nat. Protoc. 10, 1198–1211 (2015).
64. Sun, T. et al. Crosstalk between RNA m6A and DNA methylation regulates transposable element chromatin activation and cell fate in human pluripotent stem cells. Nat. Genet. 55, 1324–1335 (2023).
65. Dominissini, D., Moshitch-Moshkovitz, S., Salmon-Divon, M., Amariglio, N. & Rechavi, G. Transcriptome-wide mapping of N6-methyladenosine by m6A-seq based on immunocapturing and massively parallel sequencing. Nat. Protoc. 8, 176–189 (2013).
66. Wu, C. et al. Interplay of m 6 A and H3K27 trimethylation restrains inflammation during bacterial infection. Sci. Adv.6, (2020).